<<

Opinion

Balance of and

: implications for ,

, and social behaviors

1 2

Inga D. Neumann and Rainer Landgraf

1

Department of Behavioral and Molecular Neurobiology, University of Regensburg, Regensburg, Germany

2

Max Planck Institute of Psychiatry, Munich, Germany

Oxytocin and vasopressin are regulators of anxiety, ([5] for review of human data), for opposing effects of OXT

-coping, and sociality. They are released within and AVP on the fine-tuned regulation of emotional behav-

hypothalamic and limbic areas from , , ior. Specifically, OXT exerts and antidepressive

and perikarya independently of, or coordinated with, effects, whereas AVP predominantly increases anxiety-

from neurohypophysial terminals. Central oxy- and depression-related behaviors. We will therefore put

tocin exerts anxiolytic and antidepressive effects, where- forward the hypothesis that a dynamic balance between

as vasopressin tends to show anxiogenic and depressive the activities of brain OXT and AVP systems impacts upon

actions. Evidence from pharmacological and genetic hypothalamic and limbic circuitries involved in a broad

association studies confirms their involvement in indi- spectrum of emotional behaviors extending to psychopa-

vidual variation of emotional traits extending to psycho- thology.

pathology. Based on their opposing effects on emotional

behaviors, we propose that a balanced activity of both Central release patterns of OXT and AVP: coordinated

brain systems is important for appropriate and independent secretion into blood

emotional behaviors. Shifting the balance between the Following their neuronal synthesis in the hypothalamic

neuropeptide systems towards oxytocin, by positive supraoptic (SON) and paraventricular (PVN) nuclei (OXT,

social stimuli and/or psychopharmacotherapy, may help AVP), or in regions of the limbic system (AVP), both

to improve emotional behaviors and reinstate mental are centrally released to regulate neuronal

health. processes in a spatially and temporally fine-tuned manner.

As , following release from term-

Introduction inals they contribute to the synaptic mode of rapid infor-

Over the past years, substantial progress has been mation processing via hard-wired neuronal connections

achieved with respect to our neurobiological understand- [1,8]. A complementary mode of OXT and AVP release is

ing of the link between anxiety and stress-coping, on the non-synaptically from dendritic, somatic, and non-termi-

one hand, and social behaviors on the other. A dynamic nal axonal regions of the neuronal membrane [1,9]

interplay of genetic, epigenetic, and environmental factors (Figure 1). Upon diffusion to nearby or remote receptors

orchestrates both individual behavioral variations and the via the extracellular fluid (ECF) and ligand binding, the

etiology of anxiety- and depression-related disorders. De- association of the OXT (OXTR) and the AVP

spite this progress, available treatment options are far receptor (AVPR) subtypes AVPR1A and AVPR1B with

from being mechanism-based, which explains the need specific intraneuronal signaling cascades determines their

for innovative therapeutic interventions. One focus of acute or long-term effects [10–12]. Whereas the quality of

modern psychiatric research for future therapies has been neuropeptide-induced effects is primarily determined by

on neuropeptide systems, with oxytocin (OXT) and argi- localization of their receptors in distinct, particularly hy-

nine vasopressin (AVP) featuring prominently in such pothalamic and limbic, brain areas [10], local concentra-

endeavors [1–5]. The synthesis and release of OXT and tion of the neuropeptide ligand in the ECF and receptor

AVP within the brain are driven by anxiogenic, stressful, density are the major determinants of the intensity and

and notably social (both positive and negative) stimuli duration of such actions. Importantly, OXT and AVP

[1,6]. In turn, once released, both neuropeptides are key actions may partly overlap, due to >85% homology be-

regulators of anxiety-related behavior, passive versus ac- tween their receptors, and this has both physiological and

tive stress-coping as an indicator of depression-like behav- pharmacological implications [13,14].

ior, and multiple aspects of [1–4,7]. Simultaneous microdialysis and blood sampling has pro-

We here discuss experimental evidence, primarily from vided evidence for both coordinated and independent

rodents, but with complementary data from human studies release of OXT and AVP within the brain, and from neuro-

hypophysial terminals into blood (Figure 1), and these seem

Corresponding author: Neumann, I.D. ([email protected]) to be both -dependent and -specific [1,15].

Keywords: anxiety; depression; neuropeptide balance; oxytocin; social behavior;

Providing evidence for coordinated release, numerous

vasopressin

0166-2236/$ – see front matter ß 2012 Elsevier Ltd. All rights reserved. http://dx.doi.org/10.1016/j.tins.2012.08.004 Trends in Neurosciences, November 2012, Vol. 35, No. 11 649

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

Blood circulation

Blood-brain barrier ? Limbic regions ECF Brain OXTR

CSF Nasal PVN, AN OXT SON Socio- emotional ? behaviors

Key: Endogenous OXT Exogenous OXT OXTR in brain and periphery Pituitary

Blood circulation

Peripheral OXTR Labor, milk ejection

TRENDS in Neurosciences

Figure 1. The brain oxytocin (OXT) system: neuronal projections, release, receptor-mediated effects, and external application. Physiologically, OXT is secreted as a

into the bloodstream from axon terminals of magnocellular hypothalamic OXT via neurohemal contacts within the upon

(e.g., , suckling, stress). These neurons may also target brain (e.g., limbic) regions via axon collaterals [8]. In addition to release from axon terminals as a

, central release of OXT as a neuromodulator was shown to occur from dendrites and perikarya [1,9], explaining basal and stimulated levels in the

(ECF) of distinct brain regions, as well as spatially and temporally precise point-to-point signaling. Central release can occur both independently of, and

simultaneously with, peripheral secretion. Together with the regional distribution and density of OXT receptor (OXTR), the amount of locally released OXT largely

determines the activity of the brain OXT system, thus contributing to the regulation of emotional and social behaviors [7]. Brain OXT availability can be further raised by

intranasal administration of OXT; exogenous OXT reaches both the (CSF) of brain ventricles and the systemic circulation [110,111]. From CSF, synthetic

neuropeptides may readily diffuse through the ventricular ependyma into the ECF according to the concentration gradient; blood–brain barrier (BBB) transport (Box 1) may,

to some extent, augment brain neuropeptide levels in a concentration-dependent manner [1]. Although exemplified for the brain OXT system in this cartoon, there is

substantial evidence for a comparable neurobiology of the brain vasopressin (AVP) system with respect to neuronal synthesis, central release, peripheral secretion,

and external application [1]. Abbreviation: AN, accessory magnocellular nuclei.

physiological stimuli trigger both central and peripheral Accordingly, changes in neuropeptide concentrations in

OXT release, including birth, suckling, sexual activity, human plasma, saliva, or in a behavioral context

and various forms of stress, with essentially synergistic need to be interpreted with caution. Uncertainties as to the

behavioral and physiological actions of centrally (maternal site and dynamics of central release, and whether altered

behavior, sexual behavior, anxiolysis, social preference, and levels reflect causes or consequences of behavioral altera-

recognition) and peripherally (labor, milk ejection, ) tions, limit their plausibility. In contrast to the separation

released OXT, respectively [15,16]. Magnocellular OXT neu- of central from peripheral compartments by the blood–

rons projecting to the posterior pituitary, but also targeting brain barrier (BBB) (Box 1), these neuropeptides may

limbic regions via axon collaterals [8], may explain such readily diffuse between brain ECF and ventricular cere-

coordinated release. brospinal fluid (CSF) (Figure 1). Therefore, quantification

Although these findings speak in favor of plasma in the CSF provides at least a global measure of neuropep-

OXT as being a global biomarker of central OXT tide activity in the brain, and affords a more accurate

system activity, the temporal dynamics of central and reflection of central release patterns [1]. Importantly, in

peripheral release may substantially differ in a stimulus- any body fluid, questions about the reliability of neuropep-

dependent way. Moreover, various stressors have been tide measurements must be raised. Until assays are

shown to trigger OXT (and AVP) release within hypo- strictly validated and standardized to detect bioavailable

thalamic and limbic regions, whereas neuropeptide neuropeptide, interpretation of data (particularly from

secretion into blood remains virtually unchanged commercial assays without extraction) remains vague at [1,15,16]. best.

650

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

Box 1. OXT, AVP, and the blood–brain barrier (BBB) loss-of-function studies on AVP and its AVPR1A and

AVPR1B subtypes. Specifically, central or peripheral

The BBB prevents endogenous neuropeptides, such as OXT and

administration of AVPR antagonists, local antisense

AVP, from crossing in physiologically relevant amounts. OXT/AVP

plasma levels are generally lower than those in the ECF, further targeting of AVPR1A, AVPR knockout mice, and adeno-

restricting diffusion from blood to brain. The separation of central viral vector-induced AVPR1A upregulation [28,34–38]

and peripheral compartments under physiological conditions

have confirmed the anxiogenic effects of endogenous

coevolved with the functional divergence of neuropeptide effects

AVP. In a rat model of high (HAB) versus low (LAB)

in blood and brain. Indeed, there are primarily independent

anxiety-related behavior, representing natural extremes

physiological functions at peripheral (e.g., antidiuresis) and central

(socio-emotional behaviors) levels. In addition, coordinated and of trait anxiety, the AVP was identified as a candi-

possibly synergistic actions of peripheral and central neuropeptides

date gene for inborn anxiety [39]. As exemplified in this

may occur following simultaneous release into both compartments

model, as well as in early-life stress approaches [40,41],

(e.g., during birth). Thus, instead of being merely a protective

hyperactivity of the AVP system severely disturbs the

structure, the BBB contributes to both independent and fine-tuned

coordinated neuropeptide regulation. However, it should be noted neuropeptide balance, thus shifting behavior along a

that exogenous neuropeptides may reach the brain parenchyma continuum towards hyperanxiety and passive coping

through the BBB in minute, but functionally significant, amounts (as

(Figure 3). Consequently, reinstatement of the OXT–

indicated in Figure 1).

AVP balance, and of emotional behavior, could be

achieved by either loss-of-function approaches targeting

the AVP system [36] or, alternatively, by chronic OXT

OXT and AVP: anxiety and social phobia treatment, as shown in HAB rats [4].

Brain OXT and AVP are important regulators of anxiety, With respect to , the AVPR1A of the mouse

although usually in opposing directions. Once released in medial was suggested to mediate prosocial beha-

brain regions involved in stress and anxiety regulation, for viors, with an opposite, antisocial role for AVPR1B, em-

example in response to anxiogenic stimuli [1,15,16], OXT phasizing the potential utility of the AVPR1B antagonist

exerts anxiolytic effects and modulates neuronal functions SSR149415 in patients with social anxiety or social phobia

related to physiological stress responses, mainly at the [42]. However, an involvement of the OXT system cannot

levels of the PVN and amygdala [8,17–21]. Particularly entirely be excluded because SSR149415 has also been

intriguing is the reduction in emotional responsiveness shown to weakly bind to OXTR [13].

during periods of high activity of the endogenous OXT There is also general support for an anxiolytic effect of

system, such as during [22] and sexual activity OXT in humans. For example, nursing mothers with

[23,24]. Acute or chronic central administration of synthet- higher OXT levels are more likely to describe positive mood

ic OXT in rodents, thereby increasing neuropeptide avail- states and reduced anxiety [43,44]. By contrast, women

ability in the ECF (Figure 1), confirmed the anxiolytic and who were abused in childhood have lower OXT concentra-

stress-protective effects in various experimental settings tions in CSF and higher anxiety scores [45].

both in females and males [25–28]. Moreover, the brain In a plethora of human studies, intranasal adminis-

OXT system seems to be important for expression and tration of synthetic OXT is currently used to increase the

extinction, as shown in a rodent model of cued fear condi- availability of OXT in the brain ECF and, consequently,

tioning [8,29]. Behavioral data from transgenic mice lack- brain OXT system activity (Figure 1). Despite individual

ing either OXT or the OXTR provide further support for variations in OXT effectiveness [46], these studies sup-

involvement of the brain OXT system in anxiety regulation port the capacity of the neuropeptide to modulate anxi-

[14,30]. The OXTR-mediated acute anxiolytic effect of OXT ety circuitries, including reduced [47] and enhanced [48]

within the PVN requires the intracellular activation of amygdala reactivity to fearful faces in men and women,

signaling cascades, such as the mitogen-activated respectively, indicating gender-specific modulation of

kinase cascade, which may contribute to long-term behav- perceptual salience and the processing of social cues.

ioral adaptations via gene regulation [12,20]. Whereas there appears to be little effect of nasal OXT

It is worth emphasizing that OXT exerts various pro- on trait anxiety in healthy men [49], OXT was shown to

social effects [7,31] which may, in particular, contribute to attenuate anxiety and fear responses in social contexts

its anxiolytic effects in a social context. Naturally occurring [50] ([5,51,52] for review). Moreover, in patients suffer-

social approach and social preference behavior was shown ing from social anxiety disorder or , intranasal

to be strictly dependent on brain OXT in rats and mice, and OXT reduced several symptoms of social impairment

social anxiety prevents such behavior [32,33]. In a rodent [53–56].

model of social defeat-induced social phobia, central Supporting rodent studies, emotional effects of synthet-

administration of OXT reversed social avoidance and ic AVP in humans also include increased anxiety and fear

rescued social preference [33] (Figure 2). Thus, according responses. For example, autonomic and behavioral

to our hypothesis, activation of the brain OXT system, for responses to threatening faces were elevated upon intra-

example, by positive social stimuli or pharmacotherapy, nasal AVP [57]. Further, amygdala responses to similar

results in increased central OXT availability and a (local or socio-emotional stimulation were found to be associated

global) shift of the OXT–AVP balance towards the former, with genetic variations of AVPR1A [58] (Table 1). Although

thus resulting in reduced levels of general and social intranasal administration of AVP did not affect anxiety in

anxiety (Figure 3). healthy men [59], AVPR1B antagonists were shown to

In contrast to OXT, the brain AVP system mediates attenuate indices of anxiety and depression in animal

anxiogenic effects, as shown by a variety of gain- and models and depressed individuals [37,60].

651

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

* 120 *

(a) 90

60

30 Invesgaon me (s) me Invesgaon

0

Obje ct Social Vehicle OXTR-A AVPR1A-A Social preference test Key: Obje ct Social * 120 *

(b) 90

60

+ 30 Invesgaon me (s) me Invesgaon

0

Obje ct Social No defeat Defeat Defeat Social defeat Social preference test vehicle vehicle OXT

TRENDS in Neurosciences

Figure 2. Brain oxytocin (OXT) promotes social preference and reverses defeat-induced social phobia in rodents. (a) In the social preference test, social preference is

reflected by longer exploration of the small cage containing a conspecific (social stimulus) than an empty small cage (object stimulus). The naturally occurring social

preference is prevented by central administration of an OXT receptor (OXTR) antagonist (OXTR-A), but not of an arginine (AVPR) antagonist (AVPR1A-

A). (b) 30 min exposure to social defeat (20 min before social preference testing) prevents social preference and results in social avoidance in vehicle-treated rats. Social

phobia can be reversed by intracerebroventricular infusion of OXT 20 min before behavioral testing. *P < 0.01. Adapted, with permission, from [33].

OXT, AVP, and depression-like behavior but also to a depression-like phenotype, which could be

Due to the high degree of comorbidity between anxiety and normalized by long-term treatment with the antidepres-

depression disorders, common mediators are likely to un- sant paroxetine [70]. Analogously, both AVP and AVPR1A

derlie both conditions. Indeed, in addition to its anxiolytic mRNAs were found to be overexpressed, and the number of

effect, synthetic OXT was shown to shift stress-coping in AVP-expressing neurons increased in the PVN of de-

rodents towards a more active coping style, after either pressed patients [71,72]. Thus, shifting the neuropeptide

central or peripheral administration, indicating antide- balance towards OXT by inhibition of brain AVP might be

pressive-like effects ([4,61,62] for review). Further, there beneficial also in depression (Figure 3). Accordingly, mod-

is preclinical and clinical evidence that OXT may also ulators of AVPR activity are potential therapeutic tools,

contribute to the improvement of other depression-related such as the AVPR1B antagonist SSR149415 with anxio-

symptoms, including sexual dysfunctions [63,64], sleep lytic, , and stress-buffering effects

disturbances [65], and anhedonia ([4] for review). [37,42,73]. However, to date none of these drugs has

Another phenomenon possibly related to both depres- reached the market [38].

sion and central OXT is hippocampal neurogenesis, which

seems to be important for stress-coping and the buffering of Mechanisms of effects of OXT and AVP related to

depressive behavior [66]. OXT, but not AVP, was recently anxiety and depression

shown to stimulate neuronal growth and to rescue gluco- Multiple brain neurotransmitter and neuromodulator sys-

corticoid- or stress-induced suppression of neurogenesis in tems are presumed to interact at various brain levels to

the hippocampus of adult rats [67]. shape individual variations in emotionality. The mecha-

In depressed patients, evidence for an altered OXT nisms underlying anxiolytic and antidepressive effects of

system, as deduced from plasma and CSF levels, is limited OXT are likely to include interactions with monoaminergic,

and inconsistent [4,68]. Although increased OXT mRNA in particular the serotonergic, and corticotropin-releasing

expression and OXT immunoreactivity were found in post- factor (CRF) systems, both of which have been implicated in

mortem hypothalamic from depressed patients [69], anxiety disorders and depression [2,72,74]. A subpopulation

several questions remain – for example, whether such of OXTR-expressing serotonergic neurons exists within the

alterations represent causes or consequence of the disor- raphe nucleus [75]; in turn, stimulation of release

der, and whether antidepressant treatment can normalize activates hypothalamic OXT neurons [76]. Moreover, in

such changes. female rhesus monkeys, both serotonin and OXT are poten-

Similarly to anxiety, brain AVP appears to modulate tial targets of , and as such are likely to mediate its

depression-like behavior in an opposite manner to OXT, in prosocial and anxiolytic effects [77,78]. Thus, OXT-based

other words, shifting it towards passive stress-coping. therapy might be an additional option to reverse the postu-

Indeed, in the above-mentioned HAB rats, AVP overex- lated deficits in serotonergic (and possibly noradrenergic)

pression in the PVN not only contributed to hyperanxiety neurotransmission in depressed individuals, the more

652

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

(a) Studies on neuropeptide mechanisms have recently

• Genetic / epigenetic variation

been complemented by the demonstration that OXT and

• Social / physiological stimuli

AVP modulate anxiety responses and fear extinction in the

• Psychopharmacotherapy

central amygdala of rats in opposite manners, and target

distinct neuronal populations. Following local release,

OXT attenuated fear by acting on two major populations

Oxytocin of neurons of an inhibitory network, one inhibited by OXT,

Vasopressin but excited by AVP (via AVPR1A), the other being excited

by OXT but unresponsive to AVP [19,21]. These findings

Hypothalamic and limbic circuitries

suggest important functional implications of neuropeptide

balance not only at the behavioral but also neuronal net-

Anxiety work levels.

Similar mechanisms of OXT action are expected to

Active coping underlie the emotional and anti-stress effects seen in a

continuously rising number of human neuroimaging stud-

Passive coping (Depression)

ies after intranasal administration (Figures 1 and 3). For

(b) example, in OXTR risk allele carriers (rs53576A; Table 1)

who display deficits in socio-behavioral domains, altera-

Extremes in behavior at ends of the continuum

tions in hypothalamic-amygdala coupling were found [82].

• Hyper-anxiety, anxiety disorders • Hypo-anxiety Along the same lines, intranasal OXT has been shown to

• Passive stress coping, depression • Active stress coping

potently alter activation of the amygdala and its coupling

• High stress susceptibility • Stress resiliance

to brainstem regions in response to social and threatening

stimuli [48,49]. Intranasal administration of AVP has been

TRENDS in Neurosciences

found to modulate the activity and connectivity patterns

Figure 3. Hypothetical model depicting the balance in brain oxytocin (OXT) and

within prefrontal cortex-amygdala regions, circuitries that

arginine vasopressin (AVP) systems activity and its implications for behavioral

regulation from mental health to psychopathology. (a) Although brain OXT acts as are implicated in threat perception, the processing of

an endogenous anxiolytic and antidepressive neuropeptide, AVP exerts anxiogenic anxiety/fear, and in social behaviors [59].

and depression-like effects. Consequently, the balanced activities of the brain OXT

and AVP systems may impact upon individual variations in anxiety and stress-

coping style, indicative of depression-related behavior, along a continuum. The OXT, AVP, and social behaviors

brain OXT/AVP systems activity in hypothalamic and limbic regions and, thus,

According to the social brain hypothesis, the need to adapt

neuropeptide balance is determined by genetic, epigenetic, physiological, social,

behaviorally to increasing social complexity has substan-

and other environmental (risk) factors and can be further modulated by

psychopharmacotherapy (e.g., by administration of selective receptor tially contributed to the development of brain mass, cogni-

or antagonists). Shifting the OXT–AVP balance to the left, for example, by genetic

tive abilities, emotions, and language [83]. Brain OXT and

risk factors (Table 1) and/or by negative social cues early in life, resulting in

AVP, as well as their evolutionary ancestors, are major

reduced OXT system activity, is probably associated with increased anxiety-related

behavior and passive stress-coping, thus increasing the risk of psychopathology. A players in the complex orchestra shaping sociality, and this

neuropeptide imbalance can be reflected by reduced OXT and elevated AVP

impacts upon both anxiety and stress-coping [3,7,31]. Fol-

system activity, respectively, or both. Thus, shifting the OXT–AVP balance does not

lowing their central release, both OXT and AVP promote

necessarily mean reciprocal changes of both neuropeptide systems.

Pharmacologically, the neuropeptide balance may be shifted towards the right important aspects of social behavior, including social pref-

by intranasal application of synthetic OXT and increased brain OXT availability

erence (OXT) [33], maternal care, and (OXT and

(Figure 1), thus promoting the anxiolytic, anti-depressive, stress-protective, and

AVP) [84,85], sexual behavior (OXT) [63], pair-bonding in

prosocial effects of the brain OXT system. (b) Examples from animal and human

studies for extremes in behavioral phenotypes at the ends of the anxiety/stress- monogamous species (OXT and AVP) [31,86,87], social

coping continuum; the middle range represents species-specific phenotypic

cognition (OXT and AVP) [88–90], and inter-male aggres-

variation.

sion (AVP) [91,92]. It is of note in this context that the high

levels of sociability observed in rats after 3,4 methylene-

because some effects of selective serotonin reuptake inhibi- dioxymethamphetamine (ecstasy) administration were

tors (SSRI) are thought to be mediated by OXT [77]. Sup- shown to be OXT-mediated [93].

porting this view, polymorphisms in the serotonin In contrast to mostly opposite effects of OXT and AVP on

transporter and OXTR have recently been shown to anxiety and depression-related behavior, as discussed

interact in healthy men and women, thereby influencing above, social behaviors are often regulated in the same

their vulnerability for psychopathology [79]. direction, as seen, for example, in the context of pair-

In addition to AVP, hyperactivity of the brain CRF bonding in monogamous (although in a sex- and

system has been linked to both passive stress-coping in region-dependent manner) [31], maternal behavior [85],

rodents, and stress-related disorders such as depression and social memory [88,90]. However, only the brain OXT

[2,60,80]. In support, CRF and CRF receptor 1 genes are system appears to be essential for social preference and for

overexpressed in the PVN of both HAB rats [39,70] and the avoidance of social anxiety as a prerequisite for social

depressed patients [71,72]. OXT actions may partly be interaction, because AVP lacks such effects [33] (Figure 2).

mediated via effects on hypothalamic CRF neurons [25], Also, the facilitation of social fear extinction by OXT seems

which express OXTR [81]. Thus, following its activation by to be a neuropeptide-specific effect [94].

social or rewarding stimuli, endogenous OXT may contrib- Human studies confirm multiple prosocial effects of both

ute to the attenuation of anxiogenic, depressive, and stress OXT and AVP after intranasal administration in healthy

effects of CRF. subjects, as well as in patients with emotional or social

653

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

Table 1. Examples of genetic polymorphisms in genes encoding OXTR, AVPR1A, and AVP that are associated with emotional and a

social phenotypes

Gene/polymorphism Species Associated phenotypes Refs

Oxytocin receptor

rs53576 Humans Optimism, self-esteem, depression [124]

rd

(SNP in the 3 intron Social support seeking [117]

of the OXTR gene)

Empathy, stress reactivity [125]

or including

Sensitive parenting [126]

rs53576

Prosocial temperament; variations [82]

in hypothalamic, amygdala/cingulate

structure and function

Emotional loneliness [127]

Behavioral manifestations of prosociality [128]

Autism [129]

rs2254298 Humans Volume, function, and connectivity of [113,130]

rd

(SNP in the 3 intron and limbic brain regions;

of the OXTR gene) ethnically and sex-dependent effects

Susceptibility to anxiety, depression, autism [131,132]

Vasopressin receptor 1A

Length variation in tandem Voles , partner preference; modified [31]

repeats in promoter region receptor expression and distribution;

phenotypic confirmation in transgenic

mice and rats

RS1, RS3 Humans Autism, traits; differential [58]

(polymorphic microsatellite activation of amygdala

repeats near the promoter) Altruism, ; levels of AVPR1A mRNA [133]

in post-mortem hippocampi

Partner bonding [134]

Haplotypes consisting of Humans Autism [135]

RS1, RS3, and an intronic microsatellite

Vasopressin

Deletion in LAB promoter Mice (F2 panel Anxiety-related behavior [136]

from HAB Â

LAB crosses)

a

Examples were selected based on reproducibility and functional confirmation.

dysfunctions [5,57,95,96]. In this context, OXT is particu- psychopathologies [7]. Many other positive health effects

larly prominent in the processing of positive social stimuli of social support were described in animal and human

[49,52,97,98]. Opposing effects of OXT and AVP on social studies, for example on immunological and cardiovascular

recognition and socio-emotional perception have been de- functions [7,103,104]. It is noteworthy that even intense

scribed, with intranasal OXT elevating [48] and AVP social interaction of humans with their pets leads to elevated

impairing [99] mind-reading, respectively. Moreover, neu- plasma OXT [105], and this may give rise to some of the

ropeptide effects are nuanced, with a sizeable minority of beneficial effects described above.

human studies showing that OXT can even produce anti- Conversely, interrupted or lack of social interactions –

social effects under particular conditions [46]. anticipated as psychosocial stress – have been associated

with increased anxiety, especially social anxiety, or depres-

Influence of the social environment on brain OXT and sion-like behavior in rodents [33,101,106]. Indeed, psycho-

AVP activity social stressors including adverse social experiences early

Positive and rewarding social stimuli (such as mother– in life cause alterations in the OXT and AVP systems

offspring or socio-sexual interactions, and social support), [40,42,101,107–109]. Similarly, in humans, emotional ne-

and negative social experiences (such as defeat, subordina- glect or child maltreatment increase the risk for mental

tion, or interruption of maternal care early in life), differen- disorders and, under particular conditions, have also been

tially affect both neuropeptide systems. This is reflected by shown to be accompanied by lower OXT concentrations in

alterations in the expression, release, and receptor binding CSF in adulthood [45].

of OXT/AVP within limbic regions and, partly, in plasma Overall, whereas activation of the OXT system with

OXT or AVP concentrations [15,41,42,100–102]. We propose simultaneous inhibition of the AVP system might be a

that in this way the social environment may contribute to promising therapeutic option to treat anxiety disorders

the modulation of the activity of the brain OXT and AVP and depression due to mostly opposite emotional effects

systems, with positive stimuli being likely to shift the [4,54,55] (Figure 3), the impact of neuropeptide balance for

balance towards OXT (Figure 3). Indeed, rodent and human social behavior is less clear. Particularly in the case of

studies suggest that reinforcing positive social interactions unidirectional neuropeptide effects (e.g., on social cogni-

is generally beneficial for mental health, and improves tion), the situation is further complicated by potential

emotional stability and concomitantly protects against OXT/AVP (including antagonist [13]) cross-reactivity,

654

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

particularly at high dosages, due to the high extent of in socio-emotional behaviors [3]. Their complexity is fur-

receptor homology [14]. Whether this is relevant for human ther enhanced by gene–gene [79] and gene–environment

studies remains to be shown, given the low neuropeptide [117] interactions, including epigenetic modifications

dose that is likely to reach the brain compartment after shown to modify AVP expression by early-life stress in

intranasal administration [110,111]. In any case, the func- mice [41], and OXTR deficiency in autism [118]. Such

tional and structural overlap of the OXT and AVP systems modifications are particularly capable of complementing

emphasizes the complexity of the pharmacology involved association studies, linking them to an environmental

in developing neuropeptide-based selective psychophar- context [119].

macotherapies. Thus, both genetic and epigenetic variations are likely

to contribute to the activity of the brain OXT and AVP

Gender-dependent effects of OXT and AVP systems shaping individual anxiety- and depression-relat-

An important aspect of neuropeptide functions that balance ed behaviors and, consequently, the risk for psychopathol-

emotional behavior is the sexual dimorphism of OXT/AVP ogy (Figure 3).

systems, and this may underlie the higher incidence of

anxiety disorders and depression in women, and antisocial Neuropeptide balance in the regulation of emotional

behavior and autism in males [112]. upregulate behaviors

OXT synthesis within the PVN and regulate OXTR expres- The opposing effects of brain OXT and AVP systems on

sion in the amygdala via a- and b-receptor actions, anxiety and depression-related neuronal functions and

respectively. Sexually dimorphic amygdala reactivity to behaviors, as discussed above, support our hypothesis that

intranasal OXT [48], and gender-dependent impact of ge- a dynamic balance of the activities of the brain OXT and

netic variations in the OXTR upon hypothalamic and amyg- AVP systems impacts upon emotional behaviors along a

dala volume and functional coupling [82,113,114], further continuum from mental health to psychopathology

support the hypothesis of sex-dependent activity of the brain (Figure 3). Accordingly, psychiatric disorders can be con-

OXT system. sidered as extremes of quantitative dimensions at the

In contrast to OXT, AVP is mainly influenced by testos- negative end of a given continuum [120], explaining

terone via androgen, but also estrogen, receptor-mediated why, for example, pathological anxiety may evolve from

mechanisms [115]. It remains to be shown, to which extent normal anxiety [121]. We hypothesize that this shift to-

such mechanisms contribute to sexually dimorphic effects wards psychopathology is, at least in part, due to an OXT–

of intranasal AVP on human social communication and AVP imbalance determined by negative environmental, in

strategies in stressful contexts [57]. particular negative social, stimuli, in concert with genetic

and epigenetic (risk) factors. Conversely, based on findings

Genes of the OXT and AVP systems in association that the brain OXT system can be activated by social

studies stimuli, we further extend this model and hypothesize that

The data described so far, suggesting reliable OXT and AVP positive social interactions have the potential to shift the

effects on socio-emotional behaviors, stand in contrast to neuropeptide balance towards OXT, thereby attenuating

how little is currently known about candidate genes under- anxiety- and depression-related behaviors (Figure 3).

lying such behaviors and psychiatric disorders [116]. One Importantly, our hypothesis of an OXT–AVP balance

approach that can shed light on the genes involved is to primarily linked to emotional behaviors does not mean that

associate polymorphic variations, particularly single nucle- increased signaling of one neuropeptide is necessarily

otide polymorphisms (SNPs), with variations in emotional linked to reduced signaling of the other (although such

and social behaviors. However, genetic associations gener- regulatory capacity has recently been shown, both at neu-

ally raise issues related to replicability and the functional ropeptide ligand [122] and receptor [123] levels). Instead, it

effects of SNPs. It is, for instance, generally unknown how suggests that hypoactivity of OXT and hyperactivity of AVP,

polymorphic variations translate into differential expres- alone or together, may underlie a shift to the left along the

sion and availability of brain neuropeptides and their recep- behavioral continuum (Figure 3). In this case, we speculate

tors. Therefore, the selected examples presented in Table 1 that, in addition to appropriate stimulation of the endoge-

include only polymorphisms (i) that have repeatedly been nous system, combined psychopharmacotherapy of both an

confirmed to impact upon socio-emotional phenotypes, or (ii) OXTR and AVPR antagonists may have the poten-

with functional/structural correlates based on expression tial to synergistically improve psychopathological behavior.

and neuroimaging genetics approaches. Social dysfunctions are key symptoms not only of social

The neuropeptide variants that have been most exten- anxiety disorders, but also of several psychopathologies,

sively studied in their relation to behavioral traits are including major depression, post-traumatic stress disor-

located in the OXTR and AVPR genes (Table 1). For exam- ders, , and autism. Because OXT and AVP

ple, the A allele of rs53576 of the human OXTR gene (AA, AG modulate multiple aspects of both emotionality and social-

genotypes relative to G/G homozygotes) and ity, we further propose that a neuropeptide imbalance is

including this SNP confer particular risks for deficits in also likely to contribute to social deficits accompanying

socio-emotional domains. Compared with their receptors, psychopathologies.

however, less is known about behavioral implications of

genetic variations in neuropeptide genes (Table 1). Concluding remarks

Generally, single-gene associations are ultimately lim- Individual variations in anxiety- and depression-related

ited in their ability to explain large portions of variability behaviors are determined by the dynamic interplay of

655

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

4 Slattery, D.A. and Neumann, I.D. (2010) Oxytocin and major

Box 2. Outstanding questions

depressive disorder: experimental and clinical evidence for links to

Despite accumulating knowledge about the neurobiology of brain aetiology and possible treatment. Pharmaceuticals 3, 702–724

OXT and AVP systems from preclinical and clinical studies, several 5 Meyer-Lindenberg, A. et al. (2011) Oxytocin and vasopressin in the

questions remain. For example: : social neuropeptides for translational medicine. Nat.

 How do axonally versus dendritically released neuropeptides Rev. Neurosci. 12, 524–538

locally interact to specifically regulate socio-emotional behaviors? 6 Neumann, I.D. (2008) Brain oxytocin: a key regulator of emotional and

 Which social stimuli and epigenetic mechanisms, especially early social behaviours in both females and males. J. Neuroendocrinol. 20,

in life, modulate regional OXT/AVP and release 858–865

and, consequently, shift the OXT–AVP balance? 7 Neumann, I.D. (2009) The advantage of social living: brain

 Which neuropeptide receptor-mediated intracellular signaling neuropeptides mediate the beneficial consequences of sex and

cascades determine rapid and long-term OXT and AVP neuronal motherhood. Front. Neuroendocrinol. 30, 483–496

and behavioral effects? 8 Knobloch, H.S. et al. (2012) Evoked axonal oxytocin release in the

 How do individual variations in neuropeptide receptor genes central amygdala attenuates fear response. 73, 553–566

contribute to the endogenous OXT–AVP balance, and to the 9 Ludwig, M. et al. (2002) Intracellular stores regulate activity-

response of an individual to social stimuli and intranasal OXT? dependent neuropeptide release from dendrites. Nature 418, 85–89

 Which reliable biomarkers can be used, in addition to plasma/CSF 10 Gimpl, G. and Fahrenholz, F. (2001) The system:

neuropeptide levels and genetic risk factors, to assess central structure, function, and regulation. Physiol. Rev. 81, 629–683

neuropeptide activities, to diagnose an OXT–AVP imbalance, and 11 Viero, C. et al. (2010) Oxytocin: crossing the bridge between basic

to select patients for psychopharmacotherapy? science and pharmacotherapy. CNS Neurosci. Ther. 16, e138–e156

 Despite encouraging effects of intranasal administration of 12 van den Burg, E.H. and Neumann, I.D. (2011) Bridging the gap

synthetic OXT, transport routes to the brain, the extent and between GPCR activation and behaviour: oxytocin and

duration of behavioral effects, and acute or long-lasting interac- signalling in the hypothalamus. J. Mol. Neurosci. 43, 200–208

tions with the endogenous system are unknown. For example, 13 Griffante, C. et al. (2005) Selectivity of d[Cha4]AVP and SSR149415 at

does the rise in exogenous OXT trigger or instead inhibit human vasopressin and oxytocin receptors: evidence that SSR149415

endogenous neuropeptide activity? is a mixed vasopressin V1b/oxytocin . Br. J.

Pharmacol. 146, 744–751

14 Sala, M. et al. (2011) Pharmacologic rescue of impaired cognitive

theoretically unlimited combinations of underlying genet- flexibility, social deficits, increased aggression, and seizure

susceptibility in oxytocin receptor null mice: a neurobehavioral

ic, epigenetic, environmental, and social (risk) factors.

model of autism. Biol. Psychiatry 69, 875–882

Based on preclinical and clinical evidence, we propose that

15 Neumann, I.D. (2007) Stimuli and consequences of dendritic release of

each of these factors, alone or in concert, also contributes to

oxytocin within the brain. Biochem. Soc. Trans. 35, 1252–1257

individual differences in central OXT/AVP release pat- 16 Engelmann, M. et al. (2004) The hypothalamic–neurohypophysial

terns or receptor binding, thereby modulating their bal- system regulates the hypothalamic–pituitary–adrenal axis under

stress: an old concept revisited. Front. Neuroendocrinol. 25, 132–149

ance and thus, in turn, emotionality. Accordingly,

17 Neumann, I.D. et al. (2000) Brain oxytocin: differential inhibition of

neuropeptide imbalance and a shift of emotional behaviors

neuroendocrine stress responses and anxiety-related behaviour in

towards psychopathology (Figure 3) may be corrected by

virgin, pregnant and lactating rats. Neuroscience 95, 567–575

stimuli that facilitate and inhibit central release and 18 Bale, T.L. et al. (2001) CNS region-specific oxytocin receptor

actions of OXT and AVP, respectively, in a locally and expression: importance in regulation of anxiety and sex behavior.

J. Neurosci. 21, 2546–2552

temporally adequate manner. Although future studies are

19 Huber, D. et al. (2005) Vasopressin and oxytocin excite distinct

required to reveal the characteristics of appropriate sti-

neuronal populations in the central amygdala. Science 308, 245–248

muli in more detail (Box 2), present knowledge suggests 20 Blume, A. et al. (2008) Oxytocin reduces anxiety via ERK 1/2

that positive social stimuli may contribute to activation of activation: local effect within the rat hypothalamic paraventricular

nucleus Eur. J. Neurosci. 27, 1947–1956

the OXT system, thus rebalancing aberrant neuropeptide

21 Viviani, D. et al. (2011) Oxytocin selectively gates fear responses

signaling. Such therapeutic options, in combination with

through distinct outputs from the central amygdala. Science 333,

psychopharmacotherapy, seem to be a particularly prom- 104–107

ising approach for risk-allele carriers (Table 1) living in an 22 Slattery, D.A. and Neumann, I.D. (2007) No stress please!

adverse social environment. However, to further qualify Mechanisms of stress hyporesponsiveness of the maternal brain. J.

Physiol. 586, 377–385

the selection of potentially responsive patients for treat-

23 Waldherr, M. and Neumann, I.D. (2007) Centrally released oxytocin

ment, reliable biomarkers reflecting the dynamics of the

mediates mating-induced anxiolysis in male rats. Proc. Natl. Acad.

endogenous OXT–AVP balance need to be identified. Only

Sci. U.S.A. 104, 16681–16684

then can a combination of specific stimuli and intranasal 24 Nyuyki, K.D. et al. (2011) Yes, I am ready now: differential effects of

application of OXT agonists and/or AVPR antagonists paced versus unpaced mating on anxiety and central oxytocin release

in female rats. PLoS ONE 6, e23599

potentially improve emotional behaviors and overall men-

25 Windle, R.J. et al. (2004) Oxytocin attenuates stress-induced c-fos

tal health.

mRNA expression in specific regions associated with

modulation of hypothalamo-pituitary-adrenal activity. J. Neurosci.

References 24, 2974–2982

1 Landgraf, R. and Neumann, I.D. (2004) Vasopressin and oxytocin 26 Slattery, D.A. and Neumann, I.D. (2010) Chronic icv oxytocin

release within the brain: a dynamic concept of multiple and variable attenuates the pathological high anxiety state of selectively bred

modes of neuropeptide communication. Front. Neuroendocrinol. 25, Wistar rats. Neuropharmacology 58, 56–61

150–176 27 Missig, G. et al. (2011) Oxytocin reduces background anxiety in a fear-

2 de Kloet, E.R. et al. (2005) Stress and the brain: from adaptation to potentiated startle paradigm. Neuropsychopharmacology 35, 2607–

disease. Nat. Rev. Neurosci. 6, 463–475 2616

3 Insel, T.R. (2010) The challenge of translation in social neuroscience: a 28 Mak, P. et al. (2012) Modulation of anxiety behavior in the elevated

review of oxytocin, vasopressin, and affiliative behavior. Neuron 65, plus maze using peptidic oxytocin and vasopressin receptor ligands in

768–779 the rat. J. Psychopharmacol. 26, 532–542

656

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

29 Toth, I. et al. (2012) Central administration of oxytocin receptor 56 Andari, E. et al. (2010) Promoting social behavior with oxytocin in

ligands affects cued fear extinction in rats and mice in a timepoint- high-functioning disorders. Proc. Natl. Acad. Sci.

dependent manner. Psychopharmacology (Berl.) 223, 149–158 U.S.A. 107, 4389–4394

30 Amico, J.A. et al. (2004) Anxiety and stress responses in female 57 Thompson, R.R. et al. (2006) Sex-specific influences of vasopressin on

oxytocin deficient mice. J. Neuroendocrinol. 16, 319–324 human social communication. Proc. Natl. Acad. Sci. U.S.A. 103, 7889–

31 Donaldson, Z.R. and Young, L.J. (2008) Oxytocin, vasopressin, and the 7894

neurogenetics of sociality. Science 322, 900–904 58 Meyer-Lindenberg, A. et al. (2009) Genetic variants in AVPR1A linked

32 Takayanagi, Y. et al. (2005) Pervasive social deficits, but normal to autism predict amygdala activation and personality traits in

parturition, in oxytocin receptor-deficient mice. Proc. Natl. Acad. healthy humans. Mol. Psychiatry 14, 968–975

Sci. U.S.A. 102, 16096–16101 59 Zink, C.F. et al. (2010) Vasopressin modulates medial prefrontal

33 Lukas, M. et al. (2011) The neuropeptide oxytocin facilitates pro-social cortex-amygdala circuitry during emotion processing in humans. J.

behavior and prevents social avoidance in rats and mice. Neurosci. 30, 7017–7022

Neuropsychopharmacology 36, 2159–2168 60 Griebel, G. and Holsboer, F. (2012) Neuropeptide receptor ligands as

34 Pitkow, L.J. et al. (2001) Facilitation of affiliation and pair-bond drugs for psychiatric diseases: the end of the beginning? Nat. Rev.

formation by vasopressin receptor gene transfer into the ventral Drug Discov. 11, 462–478

forebrain of a monogamous . J. Neurosci. 21, 7392–7396 61 Nowakowska, E. et al. (2002) Role of neuropeptides in antidepressant

35 Ring, R.H. (2005) The central vasopressinergic system: examining the and memory improving effects of venlafaxine. Pol. J. Pharmacol. 54,

opportunities for psychiatric drug development. Curr. Pharm. Des. 11, 605–613

205–225 62 Ring, R.H. et al. (2010) Receptor and behavioral pharmacology of

36 Landgraf, R. (2006) The involvement of the vasopressin system in WAY-267464, a non-peptide oxytocin receptor agonist.

stress-related disorders. CNS Neurol. Disord. Drug Targets 5, 167– Neuropharmacology 58, 69–77

179 63 Melis, M.R. et al. (2007) Oxytocin injected into the ventral tegmental

37 Simon, N.G. et al. (2008) Vasopressin antagonists as and area induces penile and increases extracellular in

: recent developments. Recent Pat. CNS Drug Discov. the and paraventricular nucleus of the

3, 77–93 hypothalamus of male rats. Eur. J. Neurosci. 26, 1026–1035

38 Ryckmans, T. (2010) Modulation of the vasopressin system for the 64 Cantor, J.M. et al. (1999) Chronic fluoxetine inhibits sexual behavior

treatment of CNS diseases. Curr. Opin. Drug Discov. Devel. 13, 538– in the male rat: reversal with oxytocin. Psychopharmacology (Berl.)

547 144, 355–362

39 Landgraf, R. et al. (2007) Candidate genes of anxiety-related behavior 65 Lancel, M. et al. (2003) Intracerebral oxytocin modulates sleep–wake

in HAB/LAB rats and mice: focus on vasopressin and glyoxalase-I. behaviour in male rats. Regul. Pept. 114, 145–152

Neurosci. Biobehav. Rev. 31, 89–102 66 Snyder, J.S. et al. (2011) Adult hippocampal neurogenesis buffers

40 Veenema, A.H. et al. (2006) Effects of early life stress on adult male stress responses and depressive behaviour. Nature 476, 458–461

aggression and hypothalamic vasopressin and serotonin. Eur. J. 67 Leuner, B. et al. (2012) Oxytocin stimulates adult neurogenesis even

Neurosci. 24, 1711–1720 under conditions of stress and elevated glucocorticoids. Hippocampus

41 Murgatroyd, C. et al. (2010) Genes learn from stress: how infantile 22, 861–868

trauma programs us for depression. Epigenetics 5, 194–199 68 Parker, K.J. et al. (2010) Preliminary evidence that plasma oxytocin

42 Litvin, Y. et al. (2011) Effects of chronic social defeat on behavioral and levels are elevated in major depression. Psychiatry Res. 178, 359–362

neural correlates of sociality: vasopressin, oxytocin and the 69 Meynen, G. et al. (2007) Hypothalamic oxytocin mRNA expression and

vasopressinergic V1b receptor. Physiol. Behav. 103, 393–403 melancholic depression. Mol. Psychiatry 12, 118–119

43 Heinrichs, M. et al. (2001) Effects of suckling on hypothalamic– 70 Keck, M.E. et al. (2003) Reduction of hypothalamic vasopressinergic

pituitary–adrenal axis responses to psychosocial stress in hyperdrive contributes to clinically relevant behavioral and

postpartum lactating women. J. Clin. Endocrinol. Metab. 86, 4798– neuroendocrine effects of chronic paroxetine treatment in a

4804 psychopathological rat model. Neuropsychopharmacology 28, 235–

44 Carter, C.S. et al. (2001) Neuroendocrine and emotional changes in 243

the post-partum period. Prog. Brain Res. 133, 241–249 71 Wang, S.S. et al. (2008) Gene expression analysis in the human

45 Heim, C. et al. (2009) Lower CSF oxytocin concentrations in women hypothalamus in depression by laser microdissection and real-time

with a history of childhood abuse. Mol. Psychiatry 14, 954–958 PCR: the presence of multiple receptor imbalances. Mol. Psychiatry

46 Bartz, J.A. et al. (2011) Social effects of oxytocin in humans: context 13, 786–799 741

and person matter. Trends Cogn. Sci. 15, 301–309 72 Bao, A.M. and Swaab, D.F. (2010) Corticotropin-releasing

47 Kirsch, P. et al. (2005) Oxytocin modulates neural circuitry for social and arginine vasopressin in depression focus on the human

cognition and fear in humans. J. Neurosci. 25, 11489–11493 postmortem hypothalamus. Vitam. Horm. 82, 339–365

48 Domes, G. et al. (2010) Effects of intranasal oxytocin on emotional face 73 Urani, A. et al. (2011) The corticotropin-releasing factor 1 receptor

processing in women. Psychoneuroendocrinology 35, 83–93 antagonist, SSR125543, and the vasopressin 1b receptor antagonist,

49 Schulze, L. et al. (2011) Oxytocin increases recognition of masked SSR149415, prevent stress-induced cognitive impairment in mice.

emotional faces. Psychoneuroendocrinology 36, 1378–1382 Pharmacol. Biochem. Behav. 98, 425–431

50 Petrovic, P. et al. (2008) Oxytocin attenuates affective evaluations 74 McEwen, B.S. et al. (2010) The neurobiological properties of

of conditioned faces and amygdala activity. J. Neurosci. 28, 6607– tianeptine (Stablon): from monoamine hypothesis to glutamatergic

6615 modulation. Mol. Psychiatry 15, 237–249

51 Striepens, N. et al. (2011) Prosocial effects of oxytocin and clinical 75 Yoshida, M. et al. (2009) Evidence that oxytocin exerts anxiolytic

evidence for its therapeutic potential. Front. Neuroendocrinol. 32, effects via oxytocin receptor expressed in serotonergic neurons in

426–450 mice. J. Neurosci. 29, 2259–2271

52 Macdonald, K. and Feifel, D. (2012) Oxytocin in schizophrenia: a 76 Javed, A. et al. (1999) D-Fenfluramine induces serotonin-mediated

review of evidence for its therapeutic effects. Acta Neuropsychiatr. Fos expression in corticotropin-releasing factor and oxytocin neurons

24, 130–146 of the hypothalamus, and serotonin-independent Fos expression in

53 Hollander, E. et al. (2007) Oxytocin increases retention of social and neurons of the amygdala. Neuroscience

cognition in autism. Biol. Psychiatry 61, 498–503 90, 851–858

54 Guastella, A.J. et al. (2009) A randomized controlled trial of intranasal 77 Emiliano, A.B. et al. (2007) The interface of oxytocin-labeled cells and

oxytocin as an adjunct to exposure therapy for social anxiety disorder. serotonin transporter-containing fibers in the hypothalamus:

Psychoneuroendocrinology 34, 917–923 a substrate for SSRIs therapeutic effects? Neuropsychopharmacology

55 Labuschagne, I. et al. (2010) Oxytocin attenuates amygdala reactivity to 32, 977–988

fear in generalized social anxiety disorder. Neuropsychopharmacology 78 Michopoulos, V. et al. (2011) Estradiol effects on behavior and serum

35, 2403–2413 oxytocin are modified by social status and polymorphisms in the

657

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

serotonin transporter gene in female rhesus monkeys. Horm. Behav. 103 de Vries, A.C. et al. (2007) Social influences on stress responses and

59, 528–535 health. Psychoneuroendocrinology 32, 587–603

79 Montag, C. et al. (2011) Interaction of 5-HTTLPR and a variation on 104 Grippo, A.J. et al. (2009) Oxytocin protects against negative

the oxytocin receptor gene influences negative emotionality. Biol. behavioral and autonomic consequences of long-term social

Psychiatry 69, 601–603 isolation. Psychoneuroendocrinology 34, 1542–1553

80 Bosch, O.J. et al. (2009) The CRF system mediates increased passive 105 Nagasawa, M. et al. (2009) Dog’s gaze at its owner increases owner’s

stress-coping behavior following the loss of a bonded partner in a urinary oxytocin during social interaction. Horm. Behav. 55, 434–441

monogamous rodent. Neuropsychopharmacology 34, 1406–1415 106 Berton, O. et al. (2006) Essential role of BDNF in the mesolimbic

81 Dabrowska, J. et al. (2011) Neuroanatomical evidence for reciprocal dopamine pathway in social defeat stress. Science 311, 864–868

regulation of the corticotrophin-releasing factor and oxytocin systems 107 Francis, D.D. et al. (2002) Maternal care effects on oxytocin and

in the hypothalamus and the bed nucleus of the of the vasopressin (V1a) receptors: gender differences. J.

rat: implications for balancing stress and affect. Neuroendocrinol. 14, 349–353

Psychoneuroendocrinology 36, 1312–1326 108 Lukas, M. et al. (2010) Maternal separation interferes with

82 Tost, H. et al. (2010) A common allele in the oxytocin receptor gene developmental changes in brain vasopressin and oxytocin receptor

(OXTR) impacts prosocial temperament and human hypothalamic– binding in male rats. Neuropharmacology 58, 78–87

limbic structure and function. Proc. Natl. Acad. Sci. U.S.A. 107, 109 Timmer, M. et al. (2011) Evidence for a role of oxytocin receptors in the

13936–13941 long-term establishment of dominance hierarchies.

83 Dunbar, R.I. and Shultz, S. (2007) Evolution in the social brain. Neuropsychopharmacology 36, 2349–2356

Science 317, 1344–1347 110 Born, J. et al. (2002) Sniffing neuropeptides: a transnasal approach to

84 Numan, M. and Insel, T.R. (2003) The neurobiology of parental the human brain. Nat. Neurosci. 5, 514–516

behaviour (, Brain, and Behavior Series), Springer 111 Chang, S.W. et al. (2012) Inhaled oxytocin amplifies both vicarious

85 Bosch, O.J. and Neumann, I.D. (2008) Brain vasopressin is an reinforcement and self reinforcement in rhesus macaques (Macaca

important regulator of maternal behavior independent of dams’ mulatta). Proc. Natl. Acad. Sci. U.S.A. 109, 959–964

trait anxiety. Proc. Natl. Acad. Sci. U.S.A. 105, 17139–17144 112 Pfaff, D.W. et al. (2011) Male predominance in autism:

86 Carter, C.S. et al. (1995) Physiological substrates of mammalian neuroendocrine influences on arousal and social anxiety. Autism

monogamy: the model. Neurosci. Biobehav. Rev. 19, Res. 4, 163–176

303–314 113 Inoue, H. et al. (2010) Association between the oxytocin receptor gene

87 Jarcho, M.R. et al. (2011) Intranasal vasopressin affects pair bonding and amygdalar volume in healthy adults. Biol. Psychiatry 68, 1066–

and peripheral gene expression in male Callicebus cupreus. Genes 1072

Brain Behav. 10, 375–383 114 Furman, D.J. et al. (2011) Variant in oxytocin receptor gene is

88 Bielsky, I.F. and Young, L.J. (2004) Oxytocin, vasopressin, and social associated with amygdala volume. Psychoneuroendocrinology 36,

recognition in . 25, 1565–1574 891–897

89 Tobin, V.A. et al. (2010) An intrinsic vasopressin system in the 115 de Vries, G.J. and Soedersten, P. (2009) Sex differences in the brain:

olfactory bulb is involved in social recognition. Nature 464, 413–417 the relation between structure and function. Horm. Behav. 55, 589–

90 Gabor, C.S. et al. (2012) Interplay of oxytocin, vasopressin, and sex 596

hormones in the regulation of social recognition. Behav. Neurosci. 126, 116 Smoller, J.W. (2011) Who’s afraid of anxiety genetics? Biol. Psychiatry

97–109 69, 506–507

91 Ferris, C.F. (2008) Functional magnetic resonance imaging and the 117 Kim, H.S. et al. (2010) Culture, distress, and oxytocin receptor

neurobiology of vasopressin and oxytocin. Prog. Brain Res. 170, 305– polymorphism (OXTR) interact to influence emotional support

320 seeking. Proc. Natl. Acad. Sci. U.S.A. 107, 15717–15721

92 Neumann, I.D. et al. (2010) Aggression and anxiety: social context and 118 Gregory, S.G. et al. (2009) Genomic and epigenetic evidence for

neurobiological links. Front. Behav. Neurosci. 4, 12 oxytocin receptor deficiency in autism. BMC Med. 7, 62

93 Thompson, M.R. et al. (2007) A role for oxytocin and 5-HT1A receptors 119 Meaney, M.J. (2010) Epigenetics and the biological definition of gene

in the prosocial effects of 3,4 methylenedioxymethamphetamine  environment interactions. Child Dev. 81, 41–79

(‘ecstasy’). Neuroscience 146, 509–514 120 Plomin, R. et al. (2009) Common disorders are quantitative traits.

94 Toth, I. et al. (2012) Social fear conditioning: a novel and specific Nat. Rev. Genet. 10, 872–878

animal model to study social anxiety disorder. 121 Rosen, J.B. and Schulkin, J. (1998) From normal fear to pathological

Neuropsychopharmacology 37, 1433–1443 anxiety. Psychol. Rev. 105, 325–350

95 Campbell, A. (2010) Oxytocin and human social behavior. Pers. Soc. 122 Zelena, D. et al. (2009) Vasopressin administration into the

Psychol. Rev. 14, 281–295 paraventricular nucleus normalizes plasma oxytocin and

96 Dai, L. et al. (2012) Oxytocin and vasopressin are dysregulated in corticosterone levels in Brattleboro rats. Endocrinology 150, 2791–

Williams syndrome, a genetic disorder affecting social behavior. PLoS 2798

ONE 7, e38513 123 Nakamura, K. et al. (2008) Effects of vasopressin V1b receptor

97 Di Simplicio, M. et al. (2009) Oxytocin enhances processing of positive deficiency on adrenocorticotropin release from

versus negative emotional information in healthy male volunteers. J. cells in response to oxytocin stimulation. Endocrinology 149, 4883–

Psychopharmacol. 23, 241–248 4891

98 Riem, M.M. et al. (2012) No laughing matter: intranasal oxytocin 124 Saphire-Bernstein, S. et al. (2011) Oxytocin receptor gene (OXTR) is

administration changes functional brain connectivity during related to psychological resources. Proc. Natl. Acad. Sci. U.S.A. 108,

exposure to laughter. Neuropsychopharmacology 37, 1257– 15118–15122

1266 125 Rodrigues, S.M. et al. (2009) Oxytocin receptor genetic variation

99 Uzefovsky, F. et al. (2011) Vasopressin selectively impairs emotion relates to and stress reactivity in humans. Proc. Natl.

recognition in men. Psychoneuroendocrinology 37, 576–580 Acad. Sci. U.S.A. 106, 21437–21441

100 Veenema, A.H. et al. (2010) Distinct correlations of vasopressin 126 Bakermans-Kranenburg, M.J. and van Ijzendoorn, M.H. (2008)

release within the lateral septum and the bed nucleus of the stria Oxytocin receptor (OXTR) and serotonin transporter (5-HTT) genes

terminalis with the display of intermale aggression. Horm. Behav. 58, associated with observed parenting. Soc. Cogn. Affect. Neurosci. 3,

273–281 128–134

101 Reber, S.O. and Neumann, I.D. (2008) Defensive behavioral strategies 127 Lucht, M.J. et al. (2009) Associations between the oxytocin receptor

and enhanced state anxiety during chronic subordinate colony gene (OXTR) and affect, loneliness and intelligence in normal

housing are accompanied by reduced hypothalamic vasopressin, subjects. Prog. Neuropsychopharmacol. Biol. Psychiatry 33, 860–866

but not oxytocin, expression. Ann. N. Y. Acad. Sci. 1148, 184–195 128 Kogan, A. et al. (2011) Thin-slicing study of the oxytocin receptor

102 Gouin, J.P. et al. (2012) Plasma vasopressin and interpersonal (OXTR) gene and the evaluation and expression of the prosocial

functioning. Biol. Psychol. 91, 270–274 disposition. Proc. Natl. Acad. Sci. U.S.A. 108, 19189–19192

658

Opinion Trends in Neurosciences November 2012, Vol. 35, No. 11

129 Wermter, A.K. et al. (2010) Evidence for the involvement of genetic 133 Knafo, A. et al. (2008) Individual differences in allocation of funds

variation in the oxytocin receptor gene (OXTR) in the etiology of in the dictator game associated with length of the arginine

autistic disorders on high-functioning level. Am. J. Med. Genet. B: vasopressin 1a receptor RS3 promoter region and correlation

Neuropsychiatr. Genet. 153B, 629–639 between RS3 length and hippocampal mRNA. Genes Brain

130 Tost, H. et al. (2011) Neurogenetic effects of OXTR rs2254298 in the Behav. 7, 266–275

extended limbic system of healthy Caucasian adults. Biol. Psychiatry 134 Walum, H. et al. (2008) Genetic variation in the vasopressin receptor

70, e37–e39 1a gene (AVPR1A) associates with pair-bonding behavior in humans.

131 Thompson, R.J. et al. (2011) Oxytocin receptor gene polymorphism Proc. Natl. Acad. Sci. U.S.A. 105, 14153–14156

(rs2254298) interacts with familial risk for psychopathology to 135 Yirmiya, N. et al. (2006) Association between the arginine

predict symptoms of depression and anxiety in adolescent girls. vasopressin 1a receptor (AVPR1a) gene and autism in a family-

Psychoneuroendocrinology 36, 144–147 based study: mediation by socialization skills. Mol. Psychiatry 11,

132 Brune, M. (2012) Does the oxytocin receptor (OXTR) polymorphism 488–494

(rs2254298) confer ‘vulnerability’ for psychopathology or ‘differential 136 Bunck, M. et al. (2009) A hypomorphic vasopressin allele prevents

susceptibility’? Insights from evolution. BMC Med. 10, 38 anxiety-related behavior. PLoS ONE 4, e5129

659